Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 155
Filtrar
1.
Front Immunol ; 15: 1381919, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38799424

RESUMEN

Introduction: CD8+T cell tolerance plays an important role in tumor escape. Recent studies have shown that CD45+ erythroid progenitor cells (CD45+EPCs) generated through splenic extramedullary erythropoiesis suppress tumor immunity. However, the mechanism underlying how CD45+EPCs mediate CD8+T cell tolerance remains incompletely understood and requires further research. Methods: In this study, the antigen-processing abilities of CD45+EPCs was verified through both in vitro and in vivo experiments. We have used the method of co-culture in vitro and adoptive transfer experiments in vivo to explore the effects of CD45+EPCs on CD8+T cell tolerance. RNA-sequencing analysis and blocking experiments were used to evaluate the role of ROS in the CD45+EPC mediated tolerance of CD8+T cells. Finally, we incorporated uric acid into the adoptive transfer experiments to rescue the CD45+EPC mediated tumor-promoting effect. Results and discussion: We found that CD45+EPCs take up soluble proteins, present antigenic epitopes on their surface, and induce antigen-specific CD8+T cell anergy. In addition, we found that CD45+EPC directly nitrates tyrosine within the TCR/CD8 complex via the production of reactive oxygen species and peroxynitrite, preventing CD8+ T cells from responding to their specific peptide antigens. Furthermore, uric acid treatment effectively abolished the immunosuppressive effects of CD45+EPCs during CD8+T cell adoptive transfer, thereby enhancing the anti-tumor efficacy. These results demonstrated that CD8+T cell tolerance in tumor-bearing mice is induced by CD45+EPCs. The results of this study have direct implications for tumor immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Células Precursoras Eritroides , Tolerancia Inmunológica , Animales , Linfocitos T CD8-positivos/inmunología , Ratones , Células Precursoras Eritroides/inmunología , Células Precursoras Eritroides/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Ratones Endogámicos C57BL , Traslado Adoptivo , Especies Reactivas de Oxígeno/metabolismo , Escape del Tumor/inmunología , Línea Celular Tumoral , Ácido Úrico
2.
Stem Cell Reports ; 16(5): 1165-1181, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33979601

RESUMEN

SARS-CoV-2 infection is associated with lower blood oxygen levels, even in patients without hypoxia requiring hospitalization. This discordance illustrates the need for a more unifying explanation as to whether SARS-CoV-2 directly or indirectly affects erythropoiesis. Here, we show significantly enriched CD71+ erythroid precursors/progenitors in the blood circulation of COVID-19 patients. We found that these cells have distinctive immunosuppressive properties. In agreement, we observed a strong negative correlation between the frequency of these cells with T and B cell proportions in COVID-19 patients. The expansion of these CD71+ erythroid precursors/progenitors was negatively correlated with the hemoglobin levels. A subpopulation of abundant erythroid cells, CD45+ CD71+ cells, co-express ACE2, TMPRSS2, CD147, and CD26, and these can be infected with SARS-CoV-2. In turn, pre-treatment of erythroid cells with dexamethasone significantly diminished ACE2/TMPRSS2 expression and subsequently reduced their infectivity with SARS-CoV-2. This provides a novel insight into the impact of SARS-CoV-2 on erythropoiesis and hypoxia seen in COVID-19 patients.


Asunto(s)
Inmunidad Adaptativa/inmunología , COVID-19/patología , Células Precursoras Eritroides/virología , Eritropoyesis/fisiología , Hemoglobinas/análisis , Oxígeno/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/inmunología , COVID-19/inmunología , Dexametasona/farmacología , Células Precursoras Eritroides/inmunología , Femenino , Humanos , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , SARS-CoV-2/inmunología , Serina Endopeptidasas/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Adulto Joven
3.
Front Immunol ; 11: 597433, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329589

RESUMEN

Newborns are highly susceptible to infectious diseases. The underlying mechanism of neonatal infection susceptibility has generally been related to their under-developed immune system. Nevertheless, this notion has recently been challenged by the discovery of the physiological abundance of immunosuppressive erythroid precursors CD71+erythroid cells (CECs) in newborn mice and human cord blood. Here, as proof of concept, we show that these cells are also abundant in the peripheral blood of human newborns. Although their frequency appears to be more variable compared to their counterparts in mice, they rapidly decline by 4 weeks of age. However, their proportion remains significantly higher in infants up to six months of age compared to older infants. We found CD45 expressing CECs, as erythroid progenitors, were the prominent source of reactive oxygen species (ROS) production in both humans and mice. Interestingly, a higher proportion of CD45+CECs was observed in the spleen versus bone marrow of neonatal mice, which was associated with a higher ROS production by splenic CECs compared to their siblings in the bone marrow. CECs from human newborns suppressed cytokine production by CD14 monocytes and T cells, which was partially abrogated by apocynin in vitro. Moreover, the depletion of CECs in neonatal mice increased the number of activated effector immune cells in their spleen and liver, which rendered them more resistant to Listeria monocytogenes infection. This was evident by a significant reduction in the bacteria load in the spleen, liver and brain of treated-mice compared to the control group, which enhanced their survival rate. Our finding highlights the immunoregulatory processes mediated by CECs in newborns. Thus, such tightly regulated immune system in newborns/infants may explain one potential mechanism for the asymptomatic or mild COVID-19 infection in this population.


Asunto(s)
Antígenos CD/inmunología , Células Precursoras Eritroides , Terapia de Inmunosupresión , Listeria monocytogenes/inmunología , Listeriosis , Receptores de Transferrina/inmunología , Animales , Animales Recién Nacidos , COVID-19/inmunología , COVID-19/patología , Células Precursoras Eritroides/inmunología , Células Precursoras Eritroides/patología , Células Precursoras Eritroides/trasplante , Femenino , Xenoinjertos , Humanos , Recién Nacido , Listeriosis/inmunología , Listeriosis/patología , Listeriosis/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , SARS-CoV-2/inmunología
4.
Exp Hematol ; 89: 43-54.e2, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32750404

RESUMEN

Steady-state erythropoiesis generates new erythrocytes at a constant rate, and it has enormous productive capacity. This production is balanced by the removal of senescent erythrocytes by macrophages in the spleen and liver. Erythroid homeostasis is highly regulated to maintain sufficient erythrocytes for efficient oxygen delivery to the tissues, while avoiding viscosity problems associated with overproduction. However, there are times when this constant production of erythrocytes is inhibited or is inadequate; at these times, erythroid output is increased to compensate for the loss of production. In some cases, increased steady-state erythropoiesis can offset the loss of erythrocytes but, in response to inflammation caused by infection or tissue damage, steady-state erythropoiesis is inhibited. To maintain homeostasis under these conditions, an alternative stress erythropoiesis pathway is activated. Emerging data suggest that the bone morphogenetic protein 4 (BMP4)-dependent stress erythropoiesis pathway is integrated into the inflammatory response and generates a bolus of new erythrocytes that maintain homeostasis until steady-state erythropoiesis can resume. In this perspective, we define the mechanisms that generate new erythrocytes when steady-state erythropoiesis is impaired and discuss experimental models to study human stress erythropoiesis.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Eritrocitos/citología , Células Precursoras Eritroides/citología , Eritropoyesis/genética , Macrófagos/citología , Estrés Fisiológico/genética , Animales , Proteína Morfogenética Ósea 4/inmunología , Senescencia Celular/inmunología , Citocinas/genética , Citocinas/inmunología , Eritrocitos/inmunología , Células Precursoras Eritroides/inmunología , Eritropoyesis/inmunología , Regulación de la Expresión Génica , Humanos , Inflamación , Hígado/citología , Hígado/inmunología , Macrófagos/inmunología , Ratones , Modelos Biológicos , Fagocitosis , Bazo/citología , Bazo/inmunología , Estrés Fisiológico/inmunología
5.
PLoS Pathog ; 16(5): e1008579, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32421753

RESUMEN

Anti-helminth responses require robust type 2 cytokine production that simultaneously promotes worm expulsion and initiates the resolution of helminth-induced wounds and hemorrhaging. However, how infection-induced changes in hematopoiesis contribute to these seemingly distinct processes remains unknown. Recent studies have suggested the existence of a hematopoietic progenitor with dual mast cell-erythrocyte potential. Nonetheless, whether and how these progenitors contribute to host protection during an active infection remains to be defined. Here, we employed single cell RNA-sequencing and identified that the metabolic enzyme, carbonic anhydrase (Car) 1 marks a predefined bone marrow-resident hematopoietic progenitor cell (HPC) population. Next, we generated a Car1-reporter mouse model and found that Car1-GFP positive progenitors represent bipotent mast cell/erythrocyte precursors. Finally, we show that Car1-expressing HPCs simultaneously support mast cell and erythrocyte responses during Trichinella spiralis infection. Collectively, these data suggest that mast cell/erythrocyte precursors are mobilized to promote type 2 cytokine responses and alleviate helminth-induced blood loss, developmentally linking these processes. Collectively, these studies reveal unappreciated hematopoietic events initiated by the host to combat helminth parasites and provide insight into the evolutionary pressure that may have shaped the developmental relationship between mast cells and erythrocytes.


Asunto(s)
Células Precursoras Eritroides/inmunología , Eritropoyesis/inmunología , Mastocitos/inmunología , Mastocitosis/inmunología , Trichinella spiralis/inmunología , Triquinelosis/inmunología , Animales , Anhidrasa Carbónica I/genética , Anhidrasa Carbónica I/inmunología , Células Precursoras Eritroides/parasitología , Células Precursoras Eritroides/patología , Femenino , Mastocitos/parasitología , Mastocitos/patología , Mastocitosis/genética , Mastocitosis/patología , Ratones , Ratones Transgénicos , Triquinelosis/genética , Triquinelosis/patología
6.
FEBS Open Bio ; 10(3): 427-433, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31977161

RESUMEN

Renal anemia in chronic kidney disease is treated with recombinant human erythropoietin (rhEPO). However, some patients with anemia do not respond well to rhEPO, emphasizing the need for a more biocompatible EPO. Differentiation protocols for hepatic lineages have been modified to enable production from human induced pluripotent stem cell (hiPSC)-derived EPO-producing cells (EPO cells). However, markers for hiPSC-EPO cells are lacking, making it difficult to purify hiPSC-EPO cells and therefore to optimize EPO production and cell counts for transplantation. To address these issues, we investigated whether CD140b and CD73 could be used as markers for hiPSC-EPO cells. We measured the expression of EPO, CD140b, and CD73 in hiPSC-EPO cells and the EPO concentration in the cell supernatant by immunohistochemistry and enzyme-linked immunosorbent assays on culture day 13, revealing that expression levels of CD140b and CD73 are correlated with the level of EPO. In addition, rates of CD140b+ CD73+ cells were observed to be correlated with the concentration of EPO. Thus, our results suggest that CD140b and CD73 may be markers for hiPSC-EPO cells.


Asunto(s)
5'-Nucleotidasa/inmunología , Células Precursoras Eritroides/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/inmunología , 5'-Nucleotidasa/metabolismo , Anemia/sangre , Anemia/metabolismo , Biomarcadores , Diferenciación Celular/efectos de los fármacos , Células Precursoras Eritroides/inmunología , Eritropoyetina/metabolismo , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/metabolismo
7.
Sci Signal ; 12(598)2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31506384

RESUMEN

Inflammation alters bone marrow hematopoiesis to favor the production of innate immune effector cells at the expense of lymphoid cells and erythrocytes. Furthermore, proinflammatory cytokines inhibit steady-state erythropoiesis, which leads to the development of anemia in diseases with chronic inflammation. Acute anemia or hypoxic stress induces stress erythropoiesis, which generates a wave of new erythrocytes to maintain erythroid homeostasis until steady-state erythropoiesis can resume. Although hypoxia-dependent signaling is a key component of stress erythropoiesis, we found that inflammation also induced stress erythropoiesis in the absence of hypoxia. Using a mouse model of sterile inflammation, we demonstrated that signaling through Toll-like receptors (TLRs) paradoxically increased the phagocytosis of erythrocytes (erythrophagocytosis) by macrophages in the spleen, which enabled expression of the heme-responsive gene encoding the transcription factor SPI-C. Increased amounts of SPI-C coupled with TLR signaling promoted the expression of Gdf15 and Bmp4, both of which encode ligands that initiate the expansion of stress erythroid progenitors (SEPs) in the spleen. Furthermore, despite their inhibition of steady-state erythropoiesis in the bone marrow, the proinflammatory cytokines TNF-α and IL-1ß promoted the expansion and differentiation of SEPs in the spleen. These data suggest that inflammatory signals induce stress erythropoiesis to maintain erythroid homeostasis when inflammation inhibits steady-state erythropoiesis.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Eritropoyesis/inmunología , Hemo/inmunología , Inflamación/inmunología , Estrés Fisiológico/inmunología , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/inmunología , Proteína Morfogenética Ósea 4/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Eritrocitos/inmunología , Eritrocitos/metabolismo , Células Precursoras Eritroides/inmunología , Células Precursoras Eritroides/metabolismo , Eritropoyesis/genética , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/inmunología , Factor 15 de Diferenciación de Crecimiento/metabolismo , Hemo/metabolismo , Inflamación/genética , Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/genética , Fagocitosis/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Bazo/inmunología , Bazo/metabolismo , Estrés Fisiológico/genética
8.
Nat Med ; 24(10): 1536-1544, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30297899

RESUMEN

Impaired immunity in patients with late-stage cancer is not limited to antitumor responses, as demonstrated by poor vaccination protection and high susceptibility to infection1-3. This has been largely attributed to chemotherapy-induced impairment of innate immunity, such as neutropenia2, whereas systemic effects of tumors on hematopoiesis and adoptive immunity remain incompletely understood. Here we observed anemia associated with severe deficiency of CD8+ T cell responses against pathogens in treatment-naive mice bearing large tumors. Specifically, we identify CD45+ erythroid progenitor cells (CD71+TER119+; EPCs) as robust immunosuppressors. CD45+ EPCs, induced by tumor growth-associated extramedullary hematopoiesis, accumulate in the spleen to become a major population, outnumbering regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). The CD45+ EPC transcriptome closely resembles that of MDSCs, and, like MDSCs, reactive oxygen species production is a major mechanism underlying CD45+ EPC-mediated immunosuppression. Similarly, an immunosuppressive CD45+ EPC population was detected in patients with cancer who have anemia. These findings identify a major population of immunosuppressive cells that likely contributes to the impaired T cell responses commonly observed in patients with advanced cancer.


Asunto(s)
Anemia/inmunología , Células Precursoras Eritroides/inmunología , Células Supresoras de Origen Mieloide/inmunología , Sarcoma Mieloide/inmunología , Anemia/genética , Anemia/patología , Animales , Antígenos CD/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Humanos , Tolerancia Inmunológica , Inmunidad Innata/genética , Antígenos Comunes de Leucocito/inmunología , Ratones , Estadificación de Neoplasias , Especies Reactivas de Oxígeno/metabolismo , Receptores de Transferrina/inmunología , Sarcoma Mieloide/metabolismo , Sarcoma Mieloide/patología , Linfocitos T Reguladores/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Transfusion ; 58(11): 2675-2682, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30180269

RESUMEN

BACKGROUND: Antibody screening in pretransfusion tests is necessary to avoid critical complications of blood transfusion. Although red blood cells (RBCs) expressing relevant alloantigen(s) have been used for serologic antibody screening, little attention has been given to the use of cell lines, in which blood group antigen gene(s) are transduced, as reagent RBCs for antibody screening. STUDY DESIGN AND METHODS: The use of an erythroid progenitor cell line for serologic tests was studied. The expression of blood group antigens of erythroid progenitor cells was analyzed by genotyping and flow cytometry. Serologic analysis including hemagglutination was performed using erythroid progenitor cells to evaluate their sensitivity for antibody detection. Overexpression of exogenous erythroid antigen by lentiviral transduction was carried out and investigated for antibody detection sensitivity. RESULTS: Erythroid progenitor cells contained a substantial amount of hemoglobin and expressed sufficient levels of blood group antigens to detect corresponding monoclonal antibodies. Furthermore, the cell line could acquire an exogenous RBC antigen after lentiviral transduction and detected corresponding monoclonal and alloantibodies with equal sensitivity to antigen-positive RBCs. CONCLUSION: Application of erythroid progenitor cell lines for screening for unexpected antibodies could be helpful in solving issues such as reagent availability associated with the conventional RBC-based assay. The genetic expandability of erythroid progenitor cell lines by gene modification techniques could lead to the development of more convenient reagent RBCs.


Asunto(s)
Eritrocitos/inmunología , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/inmunología , Isoanticuerpos/inmunología , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Línea Celular , Citometría de Flujo , Humanos , Células K562
11.
Nat Commun ; 9(1): 1828, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29739946

RESUMEN

NOTCH signaling is required for the arterial specification and formation of hematopoietic stem cells (HSCs) and lympho-myeloid progenitors in the embryonic aorta-gonad-mesonephros region and extraembryonic vasculature from a distinct lineage of vascular endothelial cells with hemogenic potential. However, the role of NOTCH signaling in hemogenic endothelium (HE) specification from human pluripotent stem cell (hPSC) has not been studied. Here, using a chemically defined hPSC differentiation system combined with the use of DLL1-Fc and DAPT to manipulate NOTCH, we discover that NOTCH activation in hPSC-derived immature HE progenitors leads to formation of CD144+CD43-CD73-DLL4+Runx1 + 23-GFP+ arterial-type HE, which requires NOTCH signaling to undergo endothelial-to-hematopoietic transition and produce definitive lympho-myeloid and erythroid cells. These findings demonstrate that NOTCH-mediated arterialization of HE is an essential prerequisite for establishing definitive lympho-myeloid program and suggest that exploring molecular pathways that lead to arterial specification may aid in vitro approaches to enhance definitive hematopoiesis from hPSCs.


Asunto(s)
Arterias/citología , Endotelio Vascular/citología , Hemangioblastos/citología , Hematopoyesis , Neovascularización Fisiológica , Células Madre Pluripotentes/citología , Receptores Notch/metabolismo , Transducción de Señal , Animales , Antígenos CD/inmunología , Arterias/metabolismo , Proteínas de Unión al Calcio , Diferenciación Celular , Línea Celular , Linaje de la Célula , Rastreo Celular/instrumentación , Técnicas de Cocultivo , Embrión de Mamíferos/citología , Endotelio Vascular/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/inmunología , Hemangioblastos/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/inmunología , Células Madre Pluripotentes/inmunología
12.
Clin Lab ; 63(2): 373-378, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-28182355

RESUMEN

BACKGROUND: Pure red cell aplasia (PRCA) is an uncommon disease which involves an almost complete absence of the erythroid lineage in bone marrow (BM) and causes severe anemia. Cases due to monoclonal gammopathy occurring in plasma cell disorder have been infrequently reported. Here we report a case of PRCA associated plasma cell disorder, especially monoclonal gammopathy of undetermined significance (MGUS). METHODS: A 55-year-old male visited the ER due to general weakness. At his initial visit he exhibited severe anemia. Mild intravascular hemolysis was suspected. For anemia evaluation, BM examination was performed. In BM aspiration, almost no erythroid precursor cells were observed. Also, plasma cells were relatively elevated, at 7.2%. Serum electrophoresis and immunofixation revealed paraproteinemia of 5.1 g/L (IgG and lambda). No hypercalcemia, renal insufficiency or lytic bone lesions were found. This unusual case showed MGUS accompanied by PCRA. We were also able to assume the erythroid cell-specific restriction due to paraprotein, because we ruled out possible causes of PRCA. RESULTS: We discovered several reported cases associated with plasma cell dyscrasia. However, most of these cases involved plasma cell myeloma, characterized by high immunoglobulin burden. Our case demonstrates that PRCA is also observed in cases with MGUS, where immunoglobulin burden is low. CONCLUSIONS: It is not yet accurately known, what parts of erythroid precursors are targeted by M-protein nor what the mechanism is. Therefore, additional research into this matter is necessary.


Asunto(s)
Gammopatía Monoclonal de Relevancia Indeterminada/complicaciones , Mieloma Múltiple/complicaciones , Aplasia Pura de Células Rojas/complicaciones , Anemia/etiología , Biomarcadores/sangre , Examen de la Médula Ósea , Transfusión de Eritrocitos , Células Precursoras Eritroides/inmunología , Humanos , Masculino , Persona de Mediana Edad , Gammopatía Monoclonal de Relevancia Indeterminada/diagnóstico , Gammopatía Monoclonal de Relevancia Indeterminada/inmunología , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/inmunología , Proteínas de Mieloma/inmunología , Aplasia Pura de Células Rojas/diagnóstico , Aplasia Pura de Células Rojas/inmunología , Aplasia Pura de Células Rojas/terapia
13.
J Clin Pathol ; 69(10): 933-7, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27484914

RESUMEN

The reaction of Hector Battifora mesothelial epitope-1 (HBME-1) antibody with scattered pronormoblasts in normal bone marrow core biopsy specimens has been reported. This study evaluated the immunohistochemical profile of HBME-1 in a panel of 52 normal, dyserythropoietic and neoplastic marrow samples. We compared the staining property of HBME-1 with that of the commonly used erythroid marker, glycophorin A (CD235a) and in each case, we semi-quantitatively evaluated the HBME-1/CD235a-positive cells ratio. In normal samples, HBME-1 labelled scattered immature erythroid precursors. In dyserythropoietic specimens, HBME-1 stained nucleated erythroid precursors in varying degrees, from pronormoblast through normoblast stages, with the highest intensity in immature forms. Overall, the cellular background of non-erythroid progenitors, erythrocytes and neoplastic cells did not react with HBME-1, except in leukaemia cases with myelodysplasia-related changes. Our study shows that HBME-1 is a useful marker to identify immature erythroid precursors and that an HBME-1/CD235a-positive cells ratio ≥10% is associated with dyserythropoiesis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Médula Ósea/patología , Células Precursoras Eritroides/metabolismo , Síndromes Mielodisplásicos/metabolismo , Anticuerpos Monoclonales/inmunología , Biomarcadores de Tumor/inmunología , Biopsia con Aguja Gruesa , Células Precursoras Eritroides/inmunología , Células Precursoras Eritroides/patología , Glicoforinas/metabolismo , Humanos , Síndromes Mielodisplásicos/diagnóstico , Estudios Retrospectivos
14.
J Pediatr Hematol Oncol ; 37(6): e375-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25929611

RESUMEN

Hemolytic disease of the newborn (HDN) arising from MNSs incompatibility is rare, with few reports of prolonged anemia and reticulocytopenia following HDN. We report the younger of 2 male siblings, both of whom had anti-M-induced HDN and anemia persisting for over a month. Peripheral reticulocytes remained inappropriately low for the degree of anemia, and they needed multiple red cell transfusions. Viral infections were ruled out. Corticosteroids were given for suspected pure red cell aplasia. Anemia and reticulocytopenia subsequently improved. Colony-forming unit erythroid assay revealed erythropoietic suppression of M antigen-positive erythroid precursor cells cultured with maternal or infant sera containing anti-M. In conclusion, maternal anti-M caused HDN and prolonged anemia by erythropoietic suppression in 2 siblings.


Asunto(s)
Anemia/etiología , Eritroblastosis Fetal/etiología , Células Precursoras Eritroides/patología , Eritropoyesis/inmunología , Inmunoglobulina M/inmunología , Isoanticuerpos/inmunología , Aplasia Pura de Células Rojas/complicaciones , Adulto , Anemia/patología , Diferenciación Celular , Ensayo de Unidades Formadoras de Colonias , Eritroblastosis Fetal/patología , Células Precursoras Eritroides/inmunología , Femenino , Humanos , Inmunoglobulina M/sangre , Lactante , Recién Nacido , Masculino , Embarazo , Pronóstico , Hermanos
15.
Vox Sang ; 108(3): 302-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25523606

RESUMEN

BACKGROUND AND OBJECTIVES: Previously, a weak phenotype Am or Bm was assumed to be caused by a reduction of A or B gene expression in bone marrow cells, but not in mucus-secreting cells. However, ABO expression has not been examined in erythroid progenitor cells of Am or Bm individuals. MATERIALS AND METHODS: We carried out in vitro erythroid differentiation of CD34(+) cells from peripheral blood of a Bm individual harbouring a 3.0-kb deletion including an erythroid cell-specific regulatory element, named the +5.8-kb site, in intron 1 of the human ABO blood group gene. RESULTS: During the in vitro differentiation of CD34(+) cells from this Bm individual into erythroid cells, B-antigens were not detectable on the cultured cells by flow cytometric analysis, and allele-specific RT-PCR consistently detected the transcripts from the O allele, but not from the B allele. Moreover, chromatin immunoprecipitation assay demonstrated that both RUNX1 and GATA-2 or GATA-1 were bound to the +5.8-kb site in cultured erythroid cells expressing ABO. CONCLUSION: It is likely that the +5.8-kb site enhances transcription from the ABO promoter in erythroid cells through binding of RUNX1 and GATA-2 or GATA-1.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/genética , Antígenos CD34/metabolismo , Células Eritroides/inmunología , Células Precursoras Eritroides/inmunología , Sistema del Grupo Sanguíneo ABO/metabolismo , Alelos , Antígenos CD34/genética , Células Cultivadas , Células Eritroides/citología , Células Precursoras Eritroides/citología , Hematopoyesis , Humanos , Regiones Promotoras Genéticas
16.
Virus Res ; 190: 8-16, 2014 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-24998884

RESUMEN

Parvovirus B19 (B19V) is a small non-enveloped DNA virus of the Parvoviridae family. It is an obligate human pathogen that preferentially replicates in erythroid progenitor cells. B19V is the causative agent of multiple erythroid-related diseases due to replication-induced cytotoxicity. Despite its strong erythroid tropism and related acute disease association, B19V has been determined to persist in many other non-erythroid tissues. This review summarizes and appraises what is known about concomitant B19V DNA persistence and non-acute viral gene expression in various, particularly non-erythroid, tissue types. The methods utilized for B19V detection are described, focusing on the discrepancies in outcomes among the employed assays. The studies where investigations focused on the impact of persistent B19V expression on cellular signaling pathways are also summarized. These studies demonstrate the expanse of the types of cells capable of in vivo B19V expression as well as the possible effect of persistent viral infection on the cellular microenvironment. Overall, these reports indicate that B19V commonly persists in a wide range of both erythroid and non-erythroid tissues, and that low-level viral gene expression can be detected in some persistently infected cells. B19V capsid RNA or proteins have been reported in bone marrow, colon, heart, liver, lymphoid, synovial, testicular, and thyroid tissues. In a sub-set of these cases, B19V capsid mRNA or proteins have been associated with increased inflammatory-related gene expression. The development of standard protocols to assay for B19V infection and expression in the context of non-erythroid, non-acute disease is warranted, and with further targeted studies, may begin to elucidate the impact of persistent B19V infection in vivo. These studies may determine the most conducive cellular environment for persistent gene expression and possible impact on disease pathogenesis.


Asunto(s)
Infecciones por Parvoviridae/inmunología , Infecciones por Parvoviridae/virología , Parvovirus B19 Humano/fisiología , Progresión de la Enfermedad , Células Precursoras Eritroides/inmunología , Células Precursoras Eritroides/virología , Humanos , Especificidad de Órganos , Infecciones por Parvoviridae/patología , Parvovirus B19 Humano/genética , Replicación Viral
17.
Vox Sang ; 106(3): 242-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24795954

RESUMEN

BACKGROUND: Kell is a complex blood group system comprising 35 antigens. Kell antigens are absent from rare red cells of the Ko (null) phenotype and expressed only weakly in the Kmod phenotype. Molecular analysis of three uncommon KEL alleles elucidated the obscure pattern of inheritance of Kell antigens in one family. MATERIALS AND METHODS: Standard serological methods were employed. All exons, flanking intronic sequence and introns 15 and 16 of KEL were sequenced from genomic DNA. cDNA was obtained from erythroid cells cultured from progenitor cells isolated from peripheral blood. RESULTS: The Kmod propositus was heterozygous for two KEL mutations: c.2107G>A, p.Gly703Arg and a synonymous mutation, c.1719C>T, in the codon for p.573Gly. Sequencing of cDNA revealed that c.1719C>T caused skipping of exon 16, resulting in a silent allele. Her KEL:3,-4 brother was heterozygous for KEL*03/04 and c.1719C/T. CONCLUSION: A synonymous mutation caused complete exon skipping, despite being located 16 bases downstream of the 3' splice site, resulting in a null KEL allele. The combined effects of two mod alleles, one responsible for KEL3 expression and the other for p.Gly703Arg, were probably responsible for an unexpected KEL:3,-4 phenotype.


Asunto(s)
Alelos , Sustitución de Aminoácidos/genética , Eliminación de Gen , Sistema del Grupo Sanguíneo de Kell/genética , Sistema del Grupo Sanguíneo de Kell/inmunología , Arginina/genética , Secuencia de Bases , Células Cultivadas , Eritrocitos/inmunología , Células Precursoras Eritroides/inmunología , Femenino , Tamización de Portadores Genéticos , Glicina/genética , Humanos , Inmunofenotipificación , Sistema del Grupo Sanguíneo de Kell/sangre , Masculino , Mutación , Linaje , Sitios de Empalme de ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
Exp Mol Pathol ; 95(3): 364-75, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24184702

RESUMEN

We performed a series of experiments under a working hypothesis that cross-linked oligomers of ribosomal protein S19 (RP S19) play an essential role in definitive erythropoiesis as a ligand of the C5a receptor of erythroblasts and macrophages. We found molecules functionally and immunologically indistinguishable from RP S19 oligomers in the extracellular fluid of porcine and guinea pig bone marrow. When an increased hematopoietic state was induced in guinea pigs by bloodletting, the bone marrow RP S19 oligomer concentration was concomitantly increased. However, when the RP S19 oligomers were immunologically neutralized or the C5a receptor was pharmacologically antagonized, hyper-erythropoiesis induced by bloodletting was prevented and the anemic state was retarded in guinea pigs. When the RP S19 oligomers were neutralized in mice after bloodletting, the reactive hyper proliferation of erythroblasts in the spleen was prevented. Proerythroblasts and erythroblasts prepared by bone marrow aspiration from healthy individuals were found to express significant levels of the C5a receptor and type 2 transglutaminase genes. Majority of erythroblasts in cord blood of healthy newborns bore the C5a receptor. Taken together, these results support our hypothesis.


Asunto(s)
Médula Ósea/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoyesis/fisiología , Receptor de Anafilatoxina C5a/metabolismo , Proteínas Ribosómicas/metabolismo , Adulto , Animales , Anticuerpos Neutralizantes , Venodisección , Western Blotting , Médula Ósea/inmunología , Médula Ósea/patología , Reactivos de Enlaces Cruzados , Células Precursoras Eritroides/inmunología , Células Precursoras Eritroides/patología , Líquido Extracelular/inmunología , Líquido Extracelular/metabolismo , Femenino , Sangre Fetal/inmunología , Sangre Fetal/metabolismo , Citometría de Flujo , Proteínas de Unión al GTP/metabolismo , Cobayas , Hemoglobinas/metabolismo , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Proteína Glutamina Gamma Glutamiltransferasa 2 , Multimerización de Proteína , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Proteínas Ribosómicas/química , Proteínas Ribosómicas/inmunología , Porcinos , Transglutaminasas/metabolismo
19.
Mutat Res ; 755(2): 126-34, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23792374

RESUMEN

The peripheral blood Pig-a assay has shown promise as a tool for evaluating in vivo mutagenicity. In this study five laboratories participated in a collaborative trial that evaluated the transferability and reproducibility of a rat Pig-a assay that uses a HIS49 antibody reacts with an antigen found on erythrocytes and erythroid progenitors. In preliminary work, flow cytometry methods were established that enabled all laboratories to detect CD59-negative erythrocyte frequencies (Pig-a mutant frequencies) of <10×10(-6) in control rats. Four of the laboratories (the in-life labs) then treated male rats with a single oral dose of N-nitroso-N-ethylurea, 7,12-dimethylbenz[a]anthracene (DMBA), or 4-nitroquinoline-1-oxide (4NQO). Blood samples were collected up to 4 weeks after the treatments and analyzed by flow cytometry for the frequency of CD59-negative cells among total red blood cells (RBCs; RBC Pig-a assay). RBC Pig-a assays were conducted in the four in-life laboratories, plus a fifth laboratory that received blood samples from the other laboratories. In addition, three of the five laboratories performed a Pig-a assay on reticulocytes (RETs; PIGRET assay), using blood from the rats treated with DMBA and 4NQO. The four in-life laboratories detected consistent, time- and dose-related increases in RBC Pig-a mutant frequency (MF) for all three test articles. Furthermore, comparable results were obtained in the fifth laboratory that received blood samples from other laboratories. The three laboratories conducting the PIGRET assay also detected consistent, time- and dose-related increases in Pig-a MF, with the RET MFs increasing more rapidly with time than RBC MFs. These results indicate that rat Pig-a assays using a HIS49 antibody were transferable between laboratories and that data generated by the assays were reproducible. The findings also suggest that the PIGRET assay may detect the in vivo mutagenicity of test compounds earlier than the RBC Pig-a assay.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos CD59/análisis , Membrana Eritrocítica/inmunología , Proteínas de la Membrana/genética , Pruebas de Mutagenicidad/métodos , 4-Nitroquinolina-1-Óxido , 9,10-Dimetil-1,2-benzantraceno , Animales , Antígenos CD59/inmunología , Membrana Eritrocítica/química , Eritrocitos/química , Eritrocitos/inmunología , Células Precursoras Eritroides/química , Células Precursoras Eritroides/inmunología , Etilnitrosourea , Citometría de Flujo/métodos , Glicosilfosfatidilinositoles/deficiencia , Glicosilfosfatidilinositoles/fisiología , Japón , Laboratorios , Masculino , Proteínas de la Membrana/fisiología , Ratas , Reproducibilidad de los Resultados , Reticulocitos/química , Reticulocitos/inmunología , Sensibilidad y Especificidad
20.
Blood Rev ; 27(1): 55-62, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23261066

RESUMEN

Donor lymphocyte infusion (DLI) using unstimulated leukapheresis is one of the most effective treatment strategies for patients with hematological malignancies; its graft-versus-leukemia effects make it especially effective in chronic myeloid leukemia patients who relapsed after allogeneic stem cell transplantation (allo-HSCT). However, DLI application is limited by the development of graft-versus-host disease and aplasia, and thus cannot be routinely applied for prophylaxis. Therefore, important questions remain to be answered, such as when, and whom to DLI? Recent advances enable DLI using allografts of granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells; allodepleted donor T cells; and infusions of donor-derived, ex vivo-expanded, CD8(+) cytotoxic T lymphocyte, which can decrease relapse and improve transplant outcomes. Preemptive immunotherapy of relapse was also introduced based on the determination of mixed chimerism and minimal residual disease. In this review, we summarize the latest developments in recent strategies that will affect future DLI efficacy - focusing on the disadvantages and advantages of each protocol for the treatment, preemptive therapy, and prophylaxis of relapse.


Asunto(s)
Células Precursoras Eritroides/trasplante , Enfermedad Injerto contra Huésped/terapia , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Inmunización Pasiva , Transfusión de Linfocitos , Linfocitos T/trasplante , Quimerismo , Células Precursoras Eritroides/efectos de los fármacos , Células Precursoras Eritroides/inmunología , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/patología , Factor Estimulante de Colonias de Granulocitos/farmacología , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/mortalidad , Neoplasias Hematológicas/patología , Movilización de Célula Madre Hematopoyética , Humanos , Selección de Paciente , Recurrencia , Análisis de Supervivencia , Linfocitos T/inmunología , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA